1
|
Shi K, Zhou X, Dai J, Gao Y, Gao L, Shen Y, Chen S. Integrative Transcriptomic and Metabolomic Analysis of Muscle and Liver Reveals Key Molecular Pathways Influencing Growth Traits in Zhedong White Geese. Animals (Basel) 2025; 15:1341. [PMID: 40362156 PMCID: PMC12071005 DOI: 10.3390/ani15091341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Geese (Anser cygnoides) are popular worldwide with consumers for their unique meat quality, egg production, foie gras, and goose down; however, the key genes that influence geese growth remain elusive. To explore the mechanism of geese growth, a total of 500 Zhedong White geese were raised; four high-weight (HW) and four low-weight (LW) male geese were selected to collect carcass traits and for further transcriptomic and metabolomic analysis. The body weight and average daily gain of HW geese were significantly higher than those of the LW geese (p-value < 0.05), and the yields of the liver, gizzard, glandular stomach, and pancreas showed no significant difference between the HW and the LW group (p-value > 0.05). Compared with the LW geese, 19 differentially expressed genes (DEGs) (i.e., COL11A2, COL22A1, and TF) were detected in the breast muscle from the HW geese, which were involved in the PPAR signaling pathway, adipocytokine signaling pathway, fatty acid biosynthesis, and ferroptosis. A total of 59 differential accumulation metabolites (DAMs), which influence the pathways of glutathione metabolism and vitamin B6 metabolism, were detected in the breast muscle between the HW and LW geese. In the liver, 106 DEGs (i.e., THSD4, CREB3L3, and CNST) and 202 DAMs were found in the livers of the HW and LW groups, respectively. DEGs regulated the pathways of the TGF-beta signaling pathway, pyruvate metabolism, and adipocytokine signaling pathway; DAMs were involved in pyrimidine metabolism, nitrogen metabolism, and phenylalanine metabolism. Correlation analysis between the top DEGs and DAMs revealed that in the breast muscle, the expression levels of COL11A2 and COL22A1 were positively correlated with the content of S-(2-Hydroxy-3-buten-1-yl)glutathione. In the liver, the expression of THSD4 was positively correlated with the content of 2-Hydroxyhexadecanoic acid. In addition, one DEG (LOC106049048) and four DAMs (mogrol, brassidic acid, flabelline, and L-Leucyl-L-alanine) were shared in the breast muscle and liver. These important results contribute to improving the knowledge of goose growth and exploring the effective molecular markers that could be adopted for Zhedong White goose breeding.
Collapse
Affiliation(s)
- Kai Shi
- Ningbo Academy of Agricultural Sciences, Institute of Livestock and Poultry Research, Ningbo 315040, China; (K.S.)
| | - Xiao Zhou
- Ningbo Academy of Agricultural Sciences, Institute of Livestock and Poultry Research, Ningbo 315040, China; (K.S.)
| | - Jiuli Dai
- Ningbo Academy of Agricultural Sciences, Institute of Livestock and Poultry Research, Ningbo 315040, China; (K.S.)
| | - Yuefeng Gao
- College of Applied Engineering, Henan University of Science and Technology, Sanmenxia 472000, China;
| | - Linna Gao
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - Yangyang Shen
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China
| | - Shufang Chen
- Ningbo Academy of Agricultural Sciences, Institute of Livestock and Poultry Research, Ningbo 315040, China; (K.S.)
| |
Collapse
|
2
|
Subramanian A, Nayak PK, Schilling TF. A Cold-Active Protease Tissue Dissociation Protocol for the Preservation of the Tendon Fibroblast Transcriptome. Bio Protoc 2025; 15:e5293. [PMID: 40364987 PMCID: PMC12067300 DOI: 10.21769/bioprotoc.5293] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 05/15/2025] Open
Abstract
Traditional tissue dissociation methods for bulk- and single-cell sequencing use various protease and/or collagenase combinations at temperatures ranging from 28 to 37 °C, which cause transcriptional cell stress that may alter data interpretation. Such artifacts can be reduced by dissociating cells in cold-active proteases, but few studies have shown that this improves cell-type specific transcription, particularly in tissues hypersensitive to mechanical integrity and extracellular matrix (ECM) interactions. To address this, we have dissociated zebrafish tendons and ligaments in subtilisin A at 4 °C and compared the results with 37 °C collagenase dissociation using bulk RNA sequencing. We find that high-temperature collagenase dissociation causes general cell stress in tendon fibroblasts (tenocytes) as reported in previous studies with other cell types, but also that high temperature specifically downregulates hallmark genes involved in tenocyte specification and ECM production in vivo. Our results suggest that cold-protease dissociation reduces transcriptional artifacts and increases the robustness of RNA-sequencing datasets such that they better reflect native in vivo tissue microenvironments. Key features • Utilizing a cold-active protease derived from the Himalayan soil bacterium B. licheniformis for tissue dissociation preserves cell transcriptomes, increasing data quality of downstream sequencing experiments. • This method is reproducible and requires no extra equipment for tissue agitation. • Tenocytes isolated using this method show lower stress and better preserved native expression of key tenocyte markers and ECM genes than with traditional warm-dissociation methods. • This protocol is ideal for cell types that are particularly sensitive to microenvironment signals or are embedded in extracellular matrix.
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Pavan K. Nayak
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| |
Collapse
|
3
|
Janssens J, Mangeol P, Hecker N, Partel G, Spanier KI, Ismail JN, Hulselmans GJ, Aerts S, Schnorrer F. Spatial transcriptomics in the adult Drosophila brain and body. eLife 2025; 13:RP92618. [PMID: 40100257 PMCID: PMC11919255 DOI: 10.7554/elife.92618] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Recently, we have achieved a significant milestone with the creation of the Fly Cell Atlas. This single-nuclei atlas encompasses the entire fly, covering the entire head and body, in addition to all major organs. This atlas catalogs many hundreds of cell types, of which we annotated 250. Thus, a large number of clusters remain to be fully characterized, in particular in the brain. Furthermore, by applying single-nuclei sequencing, all information about the spatial location of the cells in the body and of about possible subcellular localization of the mRNAs within these cells is lost. Spatial transcriptomics promises to tackle these issues. In a proof-of-concept study, we have here applied spatial transcriptomics using a selected gene panel to pinpoint the locations of 150 mRNA species in the adult fly. This enabled us to map unknown clusters identified in the Fly Cell Atlas to their spatial locations in the fly brain. Additionally, spatial transcriptomics discovered interesting principles of mRNA localization and transcriptional diversity within the large and crowded muscle cells that may spark future mechanistic investigations. Furthermore, we present a set of computational tools that will allow for easier integration of spatial transcriptomics and single-cell datasets.
Collapse
Affiliation(s)
- Jasper Janssens
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Pierre Mangeol
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Nikolai Hecker
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Gabriele Partel
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Katina I Spanier
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Joy N Ismail
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Gert J Hulselmans
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Stein Aerts
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| |
Collapse
|
4
|
Ham AS, Lin S, Tse A, Thürkauf M, McGowan TJ, Jörin L, Oliveri F, Rüegg MA. Single-nuclei sequencing of skeletal muscle reveals subsynaptic-specific transcripts involved in neuromuscular junction maintenance. Nat Commun 2025; 16:2220. [PMID: 40044687 PMCID: PMC11882927 DOI: 10.1038/s41467-025-57487-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
The neuromuscular junction (NMJ) is the synapse formed between motor neurons and skeletal muscle fibers. Its stability relies on the continued expression of genes in a subset of myonuclei, called NMJ myonuclei. Here, we use single-nuclei RNA-sequencing (snRNA-seq) to identify numerous NMJ-specific transcripts. To elucidate how the NMJ transcriptome is regulated, we also performed snRNA-seq on sciatic nerve transected, botulinum toxin injected, and Musk knockout muscles. The data show that NMJ gene expression is not only driven by agrin-Lrp4/MuSK signaling but is also affected by electrical activity and trophic factors other than agrin. By selecting the three NMJ genes Etv4, Lrtm1, and Pdzrn4, we further characterize novel contributors to NMJ stability and function. AAV-mediated overexpression shows that Etv4 is sufficient to upregulate the expression of -50% of the NMJ genes in non-synaptic myonuclei, while AAV-CRISPR/Cas9-mediated muscle-specific knockout of Pdzrn4 induces NMJ fragmentation. Further investigation of Pdzrn4 revealed that it localizes to the Golgi apparatus and interacts with MuSK protein. Collectively, our data provide a rich resource of NMJ transcripts, highlight the importance of ETS transcription factors at the NMJ, and suggest a novel pathway for NMJ post-translational modifications.
Collapse
Affiliation(s)
| | - Shuo Lin
- Biozentrum, University of Basel, Basel, Switzerland
| | - Alice Tse
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | - Lena Jörin
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | |
Collapse
|
5
|
Hansson KA. Multinucleation as a buffer against gene expression noise in syncytial myofibres. J Physiol 2025; 603:1013-1016. [PMID: 39865299 DOI: 10.1113/jp288218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Affiliation(s)
- Kenth-Arne Hansson
- Norwegian University College of Health Sciences, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
6
|
Sun Z, Cheng X, Wang Z, Qiao C, Qian H, Yuan T, Lv Z, Sun W, Zhang H, Liu Y, Lu Z, Lin J, Lai C, Wang Y, Yang X, Wang X, Meng J, Bao N. Single-nucleus transcriptomics reveals subsets of degenerative myonuclei after rotator cuff tear-induced muscle atrophy. Cell Prolif 2025; 58:e13763. [PMID: 39435630 PMCID: PMC11882757 DOI: 10.1111/cpr.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Rotator cuff tear (RCT) is the primary cause of shoulder pain and disability and frequently trigger muscle degeneration characterised by muscle atrophy, fatty infiltration and fibrosis. Single-nucleus RNA sequencing (snRNA-seq) was used to reveal the transcriptional changes in the supraspinatus muscle after RCT. Supraspinatus muscles were obtained from patients with habitual shoulder dislocation (n = 3) and RCT (n = 3). In response to the RCT, trajectory analysis showed progression from normal myonuclei to ANKRD1+ myonuclei, which captured atrophy-and fatty infiltration-related regulons (KLF5, KLF10, FOSL1 and BHLHE40). Transcriptomic alterations in fibro/adipogenic progenitors (FAPs) and muscle satellite cells (MuSCs) have also been studied. By predicting cell-cell interactions, we observed communication alterations between myofibers and muscle-resident cells following RCT. Our findings reveal the plasticity of muscle cells in response to RCT and offer valuable insights into the molecular mechanisms and potential therapeutic targets of RCT.
Collapse
Affiliation(s)
- Ziying Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xi Cheng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zheng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chenfeng Qiao
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Hong Qian
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Tao Yuan
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Wenshuang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Hanwen Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhihao Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chengteng Lai
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yang Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xiaojiang Yang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xingyun Wang
- Hongqiao International Institute of Medicine, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Jia Meng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| |
Collapse
|
7
|
Keenen MM, Yang L, Liang H, Farmer VJ, Worota RE, Singh R, Gladfelter AS, Coyne CB. Comparative analysis of the syncytiotrophoblast in placenta tissue and trophoblast organoids using snRNA sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.01.601571. [PMID: 39005304 PMCID: PMC11244908 DOI: 10.1101/2024.07.01.601571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The outer surface of chorionic villi in the human placenta consists of a single multinucleated cell called the syncytiotrophoblast (STB). The unique cellular ultrastructure of the STB presents challenges in deciphering its gene expression signature at the single-cell level, as the STB contains billions of nuclei in a single cell. There are many gaps in understanding the molecular mechanisms and developmental trajectories involved in STB formation and differentiation. To identify the underlying control of the STB, we performed comparative single nucleus (SN) and single cell (SC) RNA sequencing on placental tissue and tissue-derived trophoblast organoids (TOs). We found that SN RNA sequencing was essential to capture the STB population from both tissue and TOs. Differential gene expression and pseudotime analysis of TO-derived STB identified three distinct nuclear subtypes reminiscent of those recently identified in vivo . These included a juvenile nuclear population that exhibited both CTB and STB marker expression, a population enriched in genes involved in oxygen sensing, and finally a subtype enriched in transport and GTPase signaling molecules. Notably, suspension culture conditions of TOs that restore the native orientation of the STB (STB out ) showed elevated expression of canonical STB markers and pregnancy hormones, along with a greater proportion of the STB nucleus subtype specializing in transport and GTPase signaling, compared to those cultivated with an inverted STB polarity (STB in ). Gene regulatory analysis identified novel markers of STB differentiation conserved in tissue and TOs, including the chromatin remodeler RYBP, that exhibited STB-specific RNA and protein expression. CRISPR/Cas9 knockout of RYBP in STB in TOs did not impact cell-cell fusion; however, bulk RNA sequencing revealed downregulation of the pregnancy hormone CSH1 and upregulation of multiple genes associated with the oxygen-sensing STB nuclear subtype. Finally, we compared STB gene expression signatures amongst first trimester tissue, full-term tissue, and TOs, identifying many commonalities but also notable variability across each sample type. This indicates that STB gene expression is responsive to its environmental context. Our findings emphasize the utility of TOs to accurately model STB differentiation and the distinct nuclear subtypes observed in vivo , offering a versatile platform for unraveling the molecular mechanisms governing STB functions in placental biology and disease.
Collapse
|
8
|
Lee CW, Wang BYH, Wong SH, Chen YF, Cao Q, Hsiao AWT, Fung SH, Chen YF, Wu HH, Cheng PY, Chou ZH, Lee WYW, Tsui SKW, Lee OKS. Ginkgolide B increases healthspan and lifespan of female mice. NATURE AGING 2025; 5:237-258. [PMID: 39890935 DOI: 10.1038/s43587-024-00802-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 12/20/2024] [Indexed: 02/03/2025]
Abstract
Various anti-aging interventions show promise in extending lifespan, but many are ineffective or even harmful to healthspan. Ginkgolide B (GB), derived from Ginkgo biloba, reduces aging-related morbidities such as osteoporosis, yet its effects on healthspan and longevity have not been fully understood. In this study, we found that continuous oral administration of GB to female mice beginning at 20 months of age extended median survival and median lifespan by 30% and 8.5%, respectively. GB treatment also decreased tumor incidence; enhanced muscle quality, physical performance and metabolism; and reduced systemic inflammation and senescence. Single-nucleus RNA sequencing of skeletal muscle tissue showed that GB ameliorated aging-associated changes in cell type composition, signaling pathways and intercellular communication. GB reduced aging-induced Runx1+ type 2B myonuclei through the upregulation of miR-27b-3p, which suppresses Runx1 expression. Using functional analyses, we found that Runx1 promoted senescence and cell death in muscle cells. Collectively, these findings suggest the translational potential of GB to extend healthspan and lifespan and to promote healthy aging.
Collapse
Affiliation(s)
- Chien-Wei Lee
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan.
- Department of Biomedical Engineering, China Medical University, Taichung, Taiwan.
| | - Belle Yu-Hsuan Wang
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Shing Hei Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yi-Fan Chen
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Qin Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Allen Wei-Ting Hsiao
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sin-Hang Fung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu-Fan Chen
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
- Department of Biomedical Engineering, China Medical University, Taichung, Taiwan
| | - Hao-Hsiang Wu
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Po-Yu Cheng
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Zong-Han Chou
- Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Wayne Yuk-Wai Lee
- Center for Neuromusculoskeletal Restorative Medicine, CUHK InnoHK Centres, Hong Kong Science Park, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Centre of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Stephen Kwok Wing Tsui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
9
|
Ruggieri V, Scaricamazza S, Bracaglia A, D'Ercole C, Parisi C, D'Angelo P, Proietti D, Cappelletti C, Macone A, Lozanoska-Ochser B, Bouchè M, Latella L, Valle C, Ferri A, Giordani L, Madaro L. Polyamine metabolism dysregulation contributes to muscle fiber vulnerability in ALS. Cell Rep 2025; 44:115123. [PMID: 39932195 DOI: 10.1016/j.celrep.2024.115123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/11/2024] [Accepted: 12/06/2024] [Indexed: 05/02/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease causing progressive paralysis due to motor neuron degeneration with no curative therapy despite extensive biomedical research. One of the primary targets of ALS is skeletal muscle, which undergoes profound functional changes as the disease progresses. To better understand how altered innervation interferes with muscle homeostasis during disease progression, we generated a spatial transcriptomics dataset of skeletal muscle in the SOD1G93A mouse model of ALS. Using this strategy, we identified polyamine metabolism as one of the main altered pathways in affected muscle fibers. By establishing a correlation between the vulnerability of muscle fibers and the dysregulation of this metabolic pathway, we show that disrupting polyamine homeostasis causes impairments similar to those seen in ALS muscle. Finally, we show that restoration of polyamine homeostasis rescues the muscle phenotype in SOD1G93A mice, opening new perspectives for the treatment of ALS.
Collapse
Affiliation(s)
- Veronica Ruggieri
- Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of Rome, 00161 Rome, Italy; Laboratory affiliated with Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - Silvia Scaricamazza
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, 00143 Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), 00133 Rome, Italy
| | - Andrea Bracaglia
- Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of Rome, 00161 Rome, Italy; Laboratory affiliated with Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - Chiara D'Ercole
- Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of Rome, 00161 Rome, Italy; Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Cristina Parisi
- Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of Rome, 00161 Rome, Italy; Laboratory affiliated with Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - Paolo D'Angelo
- Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of Rome, 00161 Rome, Italy; Laboratory affiliated with Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - Daisy Proietti
- Cell Therapy for Myopathies Unit, Division of Neurosciences, San Raffaele Hospital, 20132 Milano, Italy
| | - Chiara Cappelletti
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Alberto Macone
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Marina Bouchè
- Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of Rome, 00161 Rome, Italy
| | - Lucia Latella
- National Research Council (CNR), Institute of Translational Pharmacology (IFT), 00133 Rome, Italy
| | - Cristiana Valle
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, 00143 Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), 00133 Rome, Italy
| | - Alberto Ferri
- Laboratories of Neurochemistry and of Molecular and Cellular Neurobiology, IRCCS, Fondazione Santa Lucia, 00143 Rome, Italy; National Research Council (CNR), Institute of Translational Pharmacology (IFT), 00133 Rome, Italy
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France.
| | - Luca Madaro
- Department of Anatomical, Histological, Forensic Sciences and Orthopedics, Sapienza University of Rome, 00161 Rome, Italy; Laboratory affiliated with Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| |
Collapse
|
10
|
Hoegsbjerg C, Møbjerg A, Yeung CYC, Schjerling P, Krogsgaard MR, Koch M, Kjaer M, von Keudell AG, Mackey AL. Fibre type differences in the organisation of mononuclear cells and myonuclei at the tips of human myofibres. J Cell Sci 2025; 138:JCS263660. [PMID: 39704232 DOI: 10.1242/jcs.263660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
The myotendinous junction (MTJ) is a weak link in the musculoskeletal system. Here, we isolated the tips of single myofibres from healthy (non-injured) human hamstring muscles for confocal microscopy (n=6) and undertook RNAscope in situ hybridisation (n=6) to gain insight into the profiles of cells and myonuclei in this region, in a fibre type manner. A marked presence of mononuclear cells was observed coating the myofibre tips (confirmed by serial block face scanning electron microscopy and cryosection immunofluorescence), with higher numbers for type I (median 29; range 16-63) than type II (16; 9-23) myofibres (P<0.05). The number of these cells expressing COL22A1 was comparable between fibre types. Myonuclear number and density gradually increased from the myofibre proper towards the tip for both fibre types (P<0.05). COL22A1 was expressed by similar proportions of myonuclei in type I (median 26%; range 13-56) and type II (19%; 3-67) myofibre tips. 70% of the COL22A1-positive nuclei in the MTJ region were myonuclei, and the remaining 30% were MTJ cells. This insight refines our fundamental understanding of the human MTJ at the cell and structural levels.
Collapse
Affiliation(s)
- Christian Hoegsbjerg
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ask Møbjerg
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ching-Yan Chloé Yeung
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter Schjerling
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Michael R Krogsgaard
- Section for Sports Traumatology M51, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Arvind G von Keudell
- Orthopaedic Trauma Section, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
- Brigham and Women's Hospital, Department of Orthopaedic Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
11
|
von Saucken VE, Windner SE, Armetta G, Baylies MK. Postsynaptic BMP signaling regulates myonuclear properties in Drosophila larval muscles. J Cell Biol 2025; 224:e202404052. [PMID: 39475469 PMCID: PMC11530350 DOI: 10.1083/jcb.202404052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/13/2024] [Accepted: 10/13/2024] [Indexed: 11/04/2024] Open
Abstract
The syncytial mammalian muscle fiber contains a heterogeneous population of (myo)nuclei. At the neuromuscular junction (NMJ), myonuclei have specialized positioning and gene expression. However, it remains unclear how myonuclei are recruited and what regulates myonuclear output at the NMJ. Here, we identify specific properties of myonuclei located near the Drosophila larval NMJ. These synaptic myonuclei have increased size in relation to their surrounding cytoplasmic domain (size scaling), increased DNA content (ploidy), and increased levels of transcription factor pMad, a readout for BMP signaling activity. Our genetic manipulations show that local BMP signaling affects muscle size, nuclear size, ploidy, and NMJ size and function. In support, RNA sequencing analysis reveals that pMad regulates genes involved in muscle growth, ploidy (i.e., E2f1), and neurotransmission. Our data suggest that muscle BMP signaling instructs synaptic myonuclear output that positively shapes the NMJ synapse. This study deepens our understanding of how myonuclear heterogeneity supports local signaling demands to fine tune cellular function and NMJ activity.
Collapse
Affiliation(s)
- Victoria E. von Saucken
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell-Rockefeller-Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
- Biochemistry, Cell and Developmental Biology, and Molecular Biology (BCMB) Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Stefanie E. Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giovanna Armetta
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary K. Baylies
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
12
|
Joshi AS, Castillo MB, Tomaz da Silva M, Vuong AT, Gunaratne PH, Darabi R, Liu Y, Kumar A. Single-nucleus transcriptomic analysis reveals the regulatory circuitry of myofiber XBP1 during regenerative myogenesis. iScience 2024; 27:111372. [PMID: 39650729 PMCID: PMC11625362 DOI: 10.1016/j.isci.2024.111372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 12/11/2024] Open
Abstract
Endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) is activated in skeletal muscle under multiple conditions. However, the role of the UPR in the regulation of muscle regeneration remains less understood. We demonstrate that gene expression of various markers of the UPR is induced in both myogenic and non-myogenic cells in regenerating muscle. Genetic ablation of X-box binding protein 1 (XBP1), a downstream target of the Inositol requiring enzyme 1α (IRE1α) arm of the UPR, in myofibers attenuates muscle regeneration in adult mice. Single nucleus RNA sequencing (snRNA-seq) analysis showed that deletion of XBP1 in myofibers perturbs proteolytic systems and mitochondrial function in myogenic cells. Trajectory analysis of snRNA-seq dataset showed that XBP1 regulates the abundance of satellite cells and the formation of new myofibers in regenerating muscle. In addition, ablation of XBP1 disrupts the composition of non-myogenic cells in injured muscle microenvironment. Collectively, our study suggests that myofiber XBP1 regulates muscle regeneration through both cell-autonomous and -non-autonomous mechanisms.
Collapse
Affiliation(s)
- Aniket S. Joshi
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Micah B. Castillo
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Meiricris Tomaz da Silva
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Anh Tuan Vuong
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Preethi H. Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Radbod Darabi
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Yu Liu
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Ashok Kumar
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| |
Collapse
|
13
|
Hüttemeister J, Rudolph F, Radke MH, Fink C, Friedrich D, Preibisch S, Falcke M, Wagner E, Lehnart SE, Gotthardt M. Visualizing sarcomere and cellular dynamics in skeletal muscle to improve cell therapies. eLife 2024; 13:e95597. [PMID: 39688479 DOI: 10.7554/elife.95597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The giant striated muscle protein titin integrates into the developing sarcomere to form a stable myofilament system that is extended as myocytes fuse. The logistics underlying myofilament assembly and disassembly have started to emerge with the possibility to follow labeled sarcomere components. Here, we generated the mCherry knock-in at titin's Z-disk to study skeletal muscle development and remodeling. We find titin's integration into the sarcomere tightly regulated and its unexpected mobility facilitating a homogeneous distribution of titin after cell fusion - an integral part of syncytium formation and maturation of skeletal muscle. In adult mCherry-titin mice, treatment of muscle injury by implantation of titin-eGFP myoblasts reveals how myocytes integrate, fuse, and contribute to the continuous myofilament system across cell boundaries. Unlike in immature primary cells, titin proteins are retained at the proximal nucleus and do not diffuse across the whole syncytium with implications for future cell-based therapies of skeletal muscle disease.
Collapse
Affiliation(s)
- Judith Hüttemeister
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Franziska Rudolph
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael H Radke
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Claudia Fink
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dhana Friedrich
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stephan Preibisch
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Martin Falcke
- Computational Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Eva Wagner
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Stephan E Lehnart
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Gotthardt
- Translational Cardiology and Functional Genomics, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
14
|
Grima-Terrén M, Campanario S, Ramírez-Pardo I, Cisneros A, Hong X, Perdiguero E, Serrano AL, Isern J, Muñoz-Cánoves P. Muscle aging and sarcopenia: The pathology, etiology, and most promising therapeutic targets. Mol Aspects Med 2024; 100:101319. [PMID: 39312874 DOI: 10.1016/j.mam.2024.101319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Sarcopenia is a progressive muscle wasting disorder that severely impacts the quality of life of elderly individuals. Although the natural aging process primarily causes sarcopenia, it can develop in response to other conditions. Because muscle function is influenced by numerous changes that occur with age, the etiology of sarcopenia remains unclear. However, recent characterizations of the aging muscle transcriptional landscape, signaling pathway disruptions, fiber and extracellular matrix compositions, systemic metabolomic and inflammatory responses, mitochondrial function, and neurological inputs offer insights and hope for future treatments. This review will discuss age-related changes in healthy muscle and our current understanding of how this can deteriorate into sarcopenia. As our elderly population continues to grow, we must understand sarcopenia and find treatments that allow individuals to maintain independence and dignity throughout an extended lifespan.
Collapse
Affiliation(s)
- Mercedes Grima-Terrén
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Silvia Campanario
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Ignacio Ramírez-Pardo
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Andrés Cisneros
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain
| | - Xiaotong Hong
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | | | - Antonio L Serrano
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Joan Isern
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA
| | - Pura Muñoz-Cánoves
- Altos Labs, San Diego Institute of Science, San Diego, CA, 92121, USA; Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, 08003, Spain.
| |
Collapse
|
15
|
Xie Z, Liu C, Sun C, Liu Y, Peng J, Meng L, Deng J, Wang Z, Yang C, Yuan Y, Xie Z. Single-Nucleus RNA Sequencing Unravels Early Mechanisms of Human Becker Muscular Dystrophy. Ann Neurol 2024; 96:1070-1085. [PMID: 39192489 DOI: 10.1002/ana.27070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE The transcriptional heterogeneity at a single-nucleus level in human Becker muscular dystrophy (BMD) dystrophic muscle has not been explored. Here, we aimed to understand the transcriptional heterogeneity associated with myonuclei, as well as other mononucleated cell types that underly BMD pathogenesis by performing single-nucleus RNA sequencing. METHODS We profiled single-nucleus transcriptional profiles of skeletal muscle samples from 7 BMD patients and 3 normal controls. RESULTS A total of 17,216 nuclei (12,879 from BMD patients and 4,337 from controls) were classified into 13 known cell types, including 9 myogenic lineages and 4 non-myogenic lineages, and 1 unclassified nuclear type according to their cell identities. Among them, type IIx myonuclei were the first to degenerate in response to dystrophin reduction. Differential expression analysis revealed that the fibro-adipogenic progenitors (FAPs) population had the largest transcriptional changes among all cell types. Sub-clustering analysis identified a significantly compositional increase in the activated FAPs (aFAPs) subpopulation in BMD muscles. Pseudotime analysis, regulon inference, and deconvolution analysis of bulk RNA-sequencing data derived from 29 BMD patients revealed that the aFAPs subpopulation, a distinctive and previously unrecognized mononuclear subtype, was profibrogenic and expanded in BMD patients. Muscle quantitative real-time polymerase chain reaction and immunofluorescence analysis confirmed that the mRNA and protein levels of the aFAPs markers including LUM, DCN, and COL1A1 in BMD patients were significantly higher than those in controls, respectively. INTERPRETATION Our results provide insights into the transcriptional diversity of human BMD muscle at a single-nucleus resolution and new potential targets for anti-fibrosis therapies in BMD. ANN NEUROL 2024;96:1070-1085.
Collapse
Affiliation(s)
- Zhihao Xie
- Department of Neurology, Peking University First Hospital, Beijing, China
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Chengyue Sun
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yilin Liu
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Jieru Peng
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lingchao Meng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhiying Xie
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
16
|
Nicolas R, Bonnin MA, Blavet C, de Lima JE, Legallais C, Duprez D. 3D-environment and muscle contraction regulate the heterogeneity of myonuclei. Skelet Muscle 2024; 14:27. [PMID: 39529179 PMCID: PMC11552141 DOI: 10.1186/s13395-024-00359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Skeletal muscle formation involves tight interactions between muscle cells and associated connective tissue fibroblasts. Every muscle displays the same type of organisation, they are innervated in the middle and attached at both extremities to tendons. Myonuclei are heterogeneous along myotubes and regionalised according to these middle and tip domains. During development, as soon as myotubes are formed, myonuclei at muscle tips facing developing tendons display their own molecular program. In addition to molecular heterogeneity, a subset of tip myonuclei has a fibroblastic origin different to the classical somitic origin, highlighting a cellular heterogeneity of myonuclei in foetal myotubes. To gain insights on the functional relevance of myonucleus heterogeneity during limb development, we used 2D culture and co-culture systems to dissociate autonomous processes (occurring in 2D-cultures) from 3D-environment of tissue development. We also assessed the role of muscle contraction in myonucleus heterogeneity in paralysed limb muscles. The regionalisation of cellular heterogeneity was not observed in 2D cell culture systems and paralyzed muscles. The molecular signature of MTJ myonuclei was lost in a dish and paralysed muscles indicating a requirement of 3D-enviroment and muscle contraction for MTJ formation. Tip genes that maintain a regionalized expression at myotube tips in cultures are linked to sarcomeres. The behaviour of regionalized markers in cultured myotubes and paralyzed muscles allows us to speculate whether the genes intervene in myogenesis, myotube attachment or MTJ formation.
Collapse
Affiliation(s)
- Rosa Nicolas
- UMR7622, Developmental Biology Laboratory, Sorbonne Université, Institut Biologie Paris Seine, CNRS, Inserm U1156, Paris, 75005, France
| | - Marie-Ange Bonnin
- UMR7622, Developmental Biology Laboratory, Sorbonne Université, Institut Biologie Paris Seine, CNRS, Inserm U1156, Paris, 75005, France
| | - Cédrine Blavet
- UMR7622, Developmental Biology Laboratory, Sorbonne Université, Institut Biologie Paris Seine, CNRS, Inserm U1156, Paris, 75005, France
| | - Joana Esteves de Lima
- UMR7622, Developmental Biology Laboratory, Sorbonne Université, Institut Biologie Paris Seine, CNRS, Inserm U1156, Paris, 75005, France
- University Paris Est Creteil, Inserm, EnvA, EFS, AP-HP, IRMB, Creteil, 94010, France
| | - Cécile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, Compiegne, 60203, France
| | - Delphine Duprez
- UMR7622, Developmental Biology Laboratory, Sorbonne Université, Institut Biologie Paris Seine, CNRS, Inserm U1156, Paris, 75005, France.
| |
Collapse
|
17
|
Sun C, Swoboda CO, Morales FM, Calvo C, Petrany MJ, Parameswaran S, VonHandorf A, Weirauch MT, Lepper C, Millay DP. Lineage tracing of nuclei in skeletal myofibers uncovers distinct transcripts and interplay between myonuclear populations. Nat Commun 2024; 15:9372. [PMID: 39477931 PMCID: PMC11526147 DOI: 10.1038/s41467-024-53510-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Multinucleated skeletal muscle cells need to acquire additional nuclei through fusion with activated skeletal muscle stem cells when responding to both developmental and adaptive growth stimuli. A fundamental question in skeletal muscle biology has been the reason underlying this need for new nuclei in cells that already harbor hundreds of nuclei. Here we utilize nuclear RNA-sequencing approaches and develop a lineage tracing strategy capable of defining the transcriptional state of recently fused nuclei and distinguishing this state from that of pre-existing nuclei. Our findings reveal the presence of conserved markers of newly fused nuclei both during development and after a hypertrophic stimulus in the adult. However, newly fused nuclei also exhibit divergent gene expression that is determined by the myogenic environment to which they fuse. Moreover, accrual of new nuclei through fusion is required for nuclei already resident in adult myofibers to mount a normal transcriptional response to a load-inducing stimulus. We propose a model of mutual regulation in the control of skeletal muscle development and adaptations, where newly fused and pre-existing myonuclear populations influence each other to maintain optimal functional growth.
Collapse
Affiliation(s)
- Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Fabian Montecino Morales
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cristofer Calvo
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sreeja Parameswaran
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew VonHandorf
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
18
|
Martínez Mir C, Pisterzi P, De Poorter I, Rilou M, van Kranenburg M, Heijs B, Alemany A, Sage F, Geijsen N. Spatial multi-omics in whole skeletal muscle reveals complex tissue architecture. Commun Biol 2024; 7:1272. [PMID: 39369093 PMCID: PMC11455876 DOI: 10.1038/s42003-024-06949-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Myofibers are large multinucleated cells that have long thought to have a rather simple organization. Single-nucleus transcriptomics, spatial transcriptomics and spatial metabolomics analysis have revealed distinct transcription profiles in myonuclei related to myofiber type. However, the use of local tissue collection or dissociation methods have obscured the spatial organization. To elucidate the full tissue architecture, we combine two spatial omics, RNA tomography and mass spectrometry imaging. This enables us to map the spatial transcriptomic, metabolomic and lipidomic organization of the whole murine tibialis anterior muscle. Our findings on heterogeneity in fiber type proportions are validated with multiplexed immunofluorescence staining in tibialis anterior, extensor digitorum longus and soleus. Our results demonstrate unexpectedly strong regionalization of gene expression, metabolic differences and variable myofiber type proportion along the proximal-distal axis. These new insights in whole-tissue level organization reconcile sometimes conflicting results coming from previous studies relying on local sampling methods.
Collapse
Affiliation(s)
- Clara Martínez Mir
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands
| | - Paola Pisterzi
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands
| | - Isabel De Poorter
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands
| | - Maria Rilou
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands
| | - Melissa van Kranenburg
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands
| | - Bram Heijs
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands
- Center of Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
- Bruker Daltonics GmbH & Co. KG, Bremen, Germany
| | - Anna Alemany
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands
| | - Fanny Sage
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands.
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands.
| | - Niels Geijsen
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, Leiden, The Netherlands.
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, The Netherlands.
| |
Collapse
|
19
|
Agca S, Domaniku-Waraich A, Bilgic SN, Sucuoglu M, Dag M, Dogan SA, Kir S. Tumour-induced alterations in single-nucleus transcriptome of atrophying muscles indicate enhanced protein degradation and reduced oxidative metabolism. J Cachexia Sarcopenia Muscle 2024; 15:1898-1914. [PMID: 39001644 PMCID: PMC11446705 DOI: 10.1002/jcsm.13540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/07/2024] [Accepted: 06/17/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Tumour-induced skeletal muscle wasting in the context of cancer cachexia is a condition with profound implications for patient survival. The loss of muscle mass is a significant clinical obstacle and is linked to reduced tolerance to chemotherapy and increased frailty. Understanding the molecular mechanisms driving muscle atrophy is crucial for the design of new therapeutics. METHODS Lewis lung carcinoma tumours were utilized to induce cachexia and muscle atrophy in mice. Single-nucleus libraries of the tibialis anterior (TA) muscle from tumour-bearing mice and their non-tumour-bearing controls were constructed using 10X Genomics applications following the manufacturer's guidelines. RNA sequencing results were analysed with Cell Ranger software and the Seurat R package. Oxygen consumption of mitochondria isolated from TA muscle was measured using an Oroboros O2k-FluoRespirometer. Mouse primary myotubes were treated with a recombinant ectodysplasin A2 (EDA-A2) protein to activate EDA-A2 receptor (EDA2R) signalling and study changes in gene expression and oxygen consumption. RESULTS Tumour-bearing mice were sacrificed while exhibiting moderate cachexia. Average TA muscle weight was reduced by 11% (P = 0.0207) in these mice. A total of 12 335 nuclei, comprising 6422 nuclei from the control group and 5892 nuclei from atrophying muscles, were studied. The analysis of single-nucleus transcriptomes identified distinct myonuclear gene signatures and a shift towards type IIb myonuclei. Muscle atrophy-related genes, including Atrogin1, MuRF1 and Eda2r, were upregulated in these myonuclei, emphasizing their crucial roles in muscle wasting. Gene set enrichment analysis demonstrated that EDA2R activation and tumour inoculation led to similar expression patterns in muscle cells, including the stimulation of nuclear factor-kappa B, Janus kinase-signal transducer and activator of transcription and transforming growth factor-beta pathways and the suppression of myogenesis and oxidative phosphorylation. Muscle oxidative metabolism was suppressed by both tumours and EDA2R activation. CONCLUSIONS This study identified tumour-induced transcriptional changes in muscle tissue at single-nucleus resolution and highlighted the negative impact of tumours on oxidative metabolism. These findings contribute to a deeper understanding of the molecular mechanisms underlying muscle wasting.
Collapse
Affiliation(s)
- Samet Agca
- Department of Molecular Biology and Genetics, Koç University, Istanbul, 34450, Turkey
| | | | - Sevval Nur Bilgic
- Department of Molecular Biology and Genetics, Koç University, Istanbul, 34450, Turkey
| | - Melis Sucuoglu
- Department of Molecular Biology and Genetics, Koç University, Istanbul, 34450, Turkey
| | - Meric Dag
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Boğaziçi University, Istanbul, Turkey
| | - Sukru Anil Dogan
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Boğaziçi University, Istanbul, Turkey
| | - Serkan Kir
- Department of Molecular Biology and Genetics, Koç University, Istanbul, 34450, Turkey
| |
Collapse
|
20
|
Furrer R, Handschin C. Molecular aspects of the exercise response and training adaptation in skeletal muscle. Free Radic Biol Med 2024; 223:53-68. [PMID: 39059515 PMCID: PMC7617583 DOI: 10.1016/j.freeradbiomed.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/13/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
Skeletal muscle plasticity enables an enormous potential to adapt to various internal and external stimuli and perturbations. Most notably, changes in contractile activity evoke a massive remodeling of biochemical, metabolic and force-generating properties. In recent years, a large number of signals, sensors, regulators and effectors have been implicated in these adaptive processes. Nevertheless, our understanding of the molecular underpinnings of training adaptation remains rudimentary. Specifically, the mechanisms that underlie signal integration, output coordination, functional redundancy and other complex traits of muscle adaptation are unknown. In fact, it is even unclear how stimulus-dependent specification is brought about in endurance or resistance exercise. In this review, we will provide an overview on the events that describe the acute perturbations in single endurance and resistance exercise bouts. Furthermore, we will provide insights into the molecular principles of long-term training adaptation. Finally, current gaps in knowledge will be identified, and strategies for a multi-omic and -cellular analyses of the molecular mechanisms of skeletal muscle plasticity that are engaged in individual, acute exercise bouts and chronic training adaptation discussed.
Collapse
Affiliation(s)
- Regula Furrer
- Biozentrum, University of Basel, Spitalstrasse 41, 4056, Basel, Switzerland.
| | - Christoph Handschin
- Biozentrum, University of Basel, Spitalstrasse 41, 4056, Basel, Switzerland.
| |
Collapse
|
21
|
Byun WS, Lee J, Baek JH. Beyond the bulk: overview and novel insights into the dynamics of muscle satellite cells during muscle regeneration. Inflamm Regen 2024; 44:39. [PMID: 39327631 PMCID: PMC11426090 DOI: 10.1186/s41232-024-00354-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Skeletal muscle possesses remarkable regenerative capabilities, fully recovering within a month following severe acute damage. Central to this process are muscle satellite cells (MuSCs), a resident population of somatic stem cells capable of self-renewal and differentiation. Despite the highly predictable course of muscle regeneration, evaluating this process has been challenging due to the heterogeneous nature of myogenic precursors and the limited insight provided by traditional markers with overlapping expression patterns. Notably, recent advancements in single-cell technologies, such as single-cell (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq), have revolutionized muscle research. These approaches allow for comprehensive profiling of individual cells, unveiling dynamic heterogeneity among myogenic precursors and their contributions to regeneration. Through single-cell transcriptome analyses, researchers gain valuable insights into cellular diversity and functional dynamics of MuSCs post-injury. This review aims to consolidate classical and new insights into the heterogeneity of myogenic precursors, including the latest discoveries from novel single-cell technologies.
Collapse
Affiliation(s)
- Woo Seok Byun
- School of Life Science, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Jinu Lee
- School of Life Science, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Jea-Hyun Baek
- School of Life Science, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea.
| |
Collapse
|
22
|
Patsalos A, Halasz L, Oleksak D, Wei X, Nagy G, Tzerpos P, Conrad T, Hammers DW, Sweeney HL, Nagy L. Spatiotemporal transcriptomic mapping of regenerative inflammation in skeletal muscle reveals a dynamic multilayered tissue architecture. J Clin Invest 2024; 134:e173858. [PMID: 39190487 PMCID: PMC11473166 DOI: 10.1172/jci173858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Tissue regeneration is orchestrated by macrophages that clear damaged cells and promote regenerative inflammation. How macrophages spatially adapt and diversify their functions to support the architectural requirements of actively regenerating tissue remains unknown. In this study, we reconstructed the dynamic trajectories of myeloid cells isolated from acutely injured and early stage dystrophic muscles. We identified divergent subsets of monocytes/macrophages and DCs and validated markers (e.g., glycoprotein NMB [GPNMB]) and transcriptional regulators associated with defined functional states. In dystrophic muscle, specialized repair-associated subsets exhibited distinct macrophage diversity and reduced DC heterogeneity. Integrating spatial transcriptomics analyses with immunofluorescence uncovered the ordered distribution of subpopulations and multilayered regenerative inflammation zones (RIZs) where distinct macrophage subsets are organized in functional zones around damaged myofibers supporting all phases of regeneration. Importantly, intermittent glucocorticoid treatment disrupted the RIZs. Our findings suggest that macrophage subtypes mediated the development of the highly ordered architecture of regenerative tissues, unveiling the principles of the structured yet dynamic nature of regenerative inflammation supporting effective tissue repair.
Collapse
Affiliation(s)
- Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Laszlo Halasz
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Darby Oleksak
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Xiaoyan Wei
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Thomas Conrad
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - David W. Hammers
- Myology Institute and Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - H. Lee Sweeney
- Myology Institute and Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Laszlo Nagy
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
23
|
Korb A, Tajbakhsh S, Comai GE. Functional specialisation and coordination of myonuclei. Biol Rev Camb Philos Soc 2024; 99:1164-1195. [PMID: 38477382 DOI: 10.1111/brv.13063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
Myofibres serve as the functional unit for locomotion, with the sarcomere as fundamental subunit. Running the entire length of this structure are hundreds of myonuclei, located at the periphery of the myofibre, juxtaposed to the plasma membrane. Myonuclear specialisation and clustering at the centre and ends of the fibre are known to be essential for muscle contraction, yet the molecular basis of this regionalisation has remained unclear. While the 'myonuclear domain hypothesis' helped explain how myonuclei can independently govern large cytoplasmic territories, novel technologies have provided granularity on the diverse transcriptional programs running simultaneously within the syncytia and added a new perspective on how myonuclei communicate. Building upon this, we explore the critical cellular and molecular sources of transcriptional and functional heterogeneity within myofibres, discussing the impact of intrinsic and extrinsic factors on myonuclear programs. This knowledge provides new insights for understanding muscle development, repair, and disease, but also opens avenues for the development of novel and precise therapeutic approaches.
Collapse
Affiliation(s)
- Amaury Korb
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, Paris, F-75015, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, Paris, F-75015, France
| | - Glenda E Comai
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, 25 rue du Dr. Roux, Institut Pasteur, Paris, F-75015, France
| |
Collapse
|
24
|
Goodman EJ, Biltz RG, Packer JM, DiSabato DJ, Swanson SP, Oliver B, Quan N, Sheridan JF, Godbout JP. Enhanced fear memory after social defeat in mice is dependent on interleukin-1 receptor signaling in glutamatergic neurons. Mol Psychiatry 2024; 29:2321-2334. [PMID: 38459193 PMCID: PMC11412902 DOI: 10.1038/s41380-024-02456-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 03/10/2024]
Abstract
Chronic stress is associated with increased anxiety, cognitive deficits, and post-traumatic stress disorder. Repeated social defeat (RSD) in mice causes long-term stress-sensitization associated with increased microglia activation, monocyte accumulation, and enhanced interleukin (IL)-1 signaling in endothelia and neurons. With stress-sensitization, mice have amplified neuronal, immune, and behavioral responses to acute stress 24 days later. This is clinically relevant as it shares key aspects with post-traumatic stress disorder. The mechanisms underlying stress-sensitization are unclear, but enhanced fear memory may be critical. The purpose of this study was to determine the influence of microglia and IL-1R1 signaling in neurons in the development of sensitization and increased fear memory after RSD. Here, RSD accelerated fear acquisition, delayed fear extinction, and increased cued-based freezing at 0.5 day. The enhancement in contextual fear memory after RSD persisted 24 days later. Next, microglia were depleted with a CSF1R antagonist prior to RSD and several parameters were assessed. Microglia depletion blocked monocyte recruitment to the brain. Nonetheless, neuronal reactivity (pCREB) and IL-1β RNA expression in the hippocampus and enhanced fear memory after RSD were microglial-independent. Because IL-1β RNA was prominent in the hippocampus after RSD even with microglia depletion, IL-1R1 mediated signaling in glutamatergic neurons was assessed using neuronal Vglut2+/IL-1R1-/- mice. RSD-induced neuronal reactivity (pCREB) in the hippocampus and enhancement in fear memory were dependent on neuronal IL-1R1 signaling. Furthermore, single-nuclei RNA sequencing (snRNAseq) showed that RSD influenced transcription in specific hippocampal neurons (DG neurons, CA2/3, CA1 neurons) associated with glutamate signaling, inflammation and synaptic plasticity, which were neuronal IL-1R1-dependent. Furthermore, snRNAseq data provided evidence that RSD increased CREB, BDNF, and calcium signaling in DG neurons in an IL-1R1-dependent manner. Collectively, increased IL-1R1-mediated signaling (monocytes/microglia independent) in glutamatergic neurons after RSD enhanced neuronal reactivity and fear memory.
Collapse
Affiliation(s)
- Ethan J Goodman
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Rebecca G Biltz
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jonathan M Packer
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Damon J DiSabato
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Samuel P Swanson
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Braeden Oliver
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Ning Quan
- Department of Biomedical Science, Brain Institute, Florida Atlantic University, Boca Raton, FL, USA
| | - John F Sheridan
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, USA.
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| | - Jonathan P Godbout
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
25
|
Chen L, Kong X, Johnston KG, Mortazavi A, Holmes TC, Tan Z, Yokomori K, Xu X. Single-cell spatial transcriptomics reveals a dystrophic trajectory following a developmental bifurcation of myoblast cell fates in facioscapulohumeral muscular dystrophy. Genome Res 2024; 34:665-679. [PMID: 38777608 PMCID: PMC11216401 DOI: 10.1101/gr.278717.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is linked to abnormal derepression of the transcription activator DUX4. This effect is localized to a low percentage of cells, requiring single-cell analysis. However, single-cell/nucleus RNA-seq cannot fully capture the transcriptome of multinucleated large myotubes. To circumvent these issues, we use multiplexed error-robust fluorescent in situ hybridization (MERFISH) spatial transcriptomics that allows profiling of RNA transcripts at a subcellular resolution. We simultaneously examined spatial distributions of 140 genes, including 24 direct DUX4 targets, in in vitro differentiated myotubes and unfused mononuclear cells (MNCs) of control, isogenic D4Z4 contraction mutant and FSHD patient samples, as well as the individual nuclei within them. We find myocyte nuclei segregate into two clusters defined by the expression of DUX4 target genes, which is exclusively found in patient/mutant nuclei, whereas MNCs cluster based on developmental states. Patient/mutant myotubes are found in "FSHD-hi" and "FSHD-lo" states with the former signified by high DUX4 target expression and decreased muscle gene expression. Pseudotime analyses reveal a clear bifurcation of myoblast differentiation into control and FSHD-hi myotube branches, with variable numbers of DUX4 target-expressing nuclei found in multinucleated FSHD-hi myotubes. Gene coexpression modules related to extracellular matrix and stress gene ontologies are significantly altered in patient/mutant myotubes compared with the control. We also identify distinct subpathways within the DUX4 gene network that may differentially contribute to the disease transcriptomic phenotype. Taken together, our MERFISH-based study provides effective gene network profiling of multinucleated cells and identifies FSHD-induced transcriptomic alterations during myoblast differentiation.
Collapse
Affiliation(s)
- Lujia Chen
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, California 92697, USA
| | - Xiangduo Kong
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California 92697, USA
| | - Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine, California 92697, USA
| | - Ali Mortazavi
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping, University of California, Irvine, California 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, California 92697, USA
| | - Zhiqun Tan
- Center for Neural Circuit Mapping, University of California, Irvine, California 92697, USA;
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California 92697, USA
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine, California 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California 92697, USA
| | - Kyoko Yokomori
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California 92697, USA;
| | - Xiangmin Xu
- Department of Biomedical Engineering, University of California, Irvine, California 92697, USA;
- Center for Neural Circuit Mapping, University of California, Irvine, California 92697, USA
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine, California 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California 92697, USA
- Department of Computer Science, University of California, Irvine, California 92697, USA
| |
Collapse
|
26
|
Kaplan MM, Zeidler M, Knapp A, Hölzl M, Kress M, Fritsch H, Krogsdam A, Flucher BE. Spatial transcriptomics in embryonic mouse diaphragm muscle reveals regional gradients and subdomains of developmental gene expression. iScience 2024; 27:110018. [PMID: 38883818 PMCID: PMC11177202 DOI: 10.1016/j.isci.2024.110018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/22/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
The murine embryonic diaphragm is a primary model for studying myogenesis and neuro-muscular synaptogenesis, both representing processes regulated by spatially organized genetic programs of myonuclei located in distinct myodomains. However, a spatial gene expression pattern of embryonic mouse diaphragm has not been reported. Here, we provide spatially resolved gene expression data for horizontally sectioned embryonic mouse diaphragms at embryonic days E14.5 and E18.5. These data reveal gene signatures for specific muscle regions with distinct maturity and fiber type composition, as well as for a central neuromuscular junction (NMJ) and a peripheral myotendinous junction (MTJ) compartment. Comparing spatial expression patterns of wild-type mice with those of transgenic mice lacking either the skeletal muscle calcium channel CaV1.1 or β-catenin, reveals curtailed muscle development and dysregulated expression of genes potentially involved in NMJ formation. Altogether, these datasets provide a powerful resource for further studies of muscle development and NMJ formation in the mouse.
Collapse
Affiliation(s)
| | - Maximilian Zeidler
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Annabella Knapp
- Institute of Clinical and Functional Anatomy, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Martina Hölzl
- Deep Sequencing Core and Institute for Bioinformatics Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Helga Fritsch
- Institute of Clinical and Functional Anatomy, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Anne Krogsdam
- Deep Sequencing Core and Institute for Bioinformatics Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Bernhard E Flucher
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
27
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
28
|
Funk OH, Levy DL, Fay DS. Epidermal cell fusion promotes the transition from an embryonic to a larval transcriptome in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595354. [PMID: 38826300 PMCID: PMC11142173 DOI: 10.1101/2024.05.22.595354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Cell fusion is a fundamental process in the development of multicellular organisms, yet its impact on gene regulation, particularly during crucial developmental stages, remains poorly understood. The Caenorhabditis elegans epidermis comprises 8-10 syncytial cells, with the largest integrating 139 individual nuclei through cell-cell fusion governed by the fusogenic protein EFF-1. To explore the effects of cell fusion on developmental progression and associated gene expression changes, we conducted transcriptomic analyses of eff-1 fusion-deficient mutants. Our RNAseq findings showed widespread transcriptomic changes that were enriched for epidermal genes and key molecular pathways involved in epidermal function during larval development. Subsequent single-molecule fluorescence in situ hybridization validated the altered expression of mRNA transcripts, confirming quantifiable changes in gene expression in the absence of embryonic epidermal fusion. These results underscore the significance of cell-cell fusion in shaping transcriptional programs during development and raise questions regarding the precise identities and specialized functions of different subclasses of nuclei within developing syncytial cells and tissues.
Collapse
|
29
|
Vemula S, Muvavarirwa T, Doornbos F, Whitman MC. Neuromuscular Junction Development Differs Between Extraocular and Skeletal Muscles and Between Different Extraocular Muscles. Invest Ophthalmol Vis Sci 2024; 65:28. [PMID: 38767908 PMCID: PMC11114620 DOI: 10.1167/iovs.65.5.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose To determine whether development of neuromuscular junctions (NMJs) differs between extraocular muscles (EOMs) and other skeletal muscles. Methods Mouse EOMs, diaphragm, and tibialis anterior (TA) were collected at postnatal day (P)0, P3, P7, P10, P14, and P21, and 12 weeks. Whole muscles were stained with α-bungarotoxin, anti-neurofilament antibody, and slow or fast myosin heavy chain antibody, and imaged with a confocal microscope. Images were quantified using Imaris software. Results NMJs in the EOMs show a unique pattern of morphological development compared to diaphragm and TA. At P0, diaphragm and TA NMJs were oval plaques; EOM single NMJs were long, thin rods. NMJs in the three muscle types progress to mature morphology at different rates. At all ages, EOM single NMJs were larger, especially relative to myofiber size. The inferior oblique and inferior rectus muscles show delayed single NMJ development compared to other EOMs. NMJs on multiply-innervated fibers in the EOMs vary widely in size, and there were no consistent differences between muscles or over time. Incoming motor nerves formed complex branching patterns, dividing first into superficial and deep branches, each of which branched extensively over the full width of the muscle. Motor axons that innervate multiply-innervated fibers entered the muscle with the axons that innervate singly-innervated fibers, then extended both proximally and distally. EOM NMJs had more subsynaptic nuclei than skeletal muscle NMJs throughout development. Conclusions EOMs show a unique pattern of NMJ development and have more subsynaptic nuclei than other muscles, which may contribute to the exquisite control of eye movements.
Collapse
Affiliation(s)
- Sampath Vemula
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Tapiwa Muvavarirwa
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Fleur Doornbos
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Mary C. Whitman
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, United States
| |
Collapse
|
30
|
Kedlian VR, Wang Y, Liu T, Chen X, Bolt L, Tudor C, Shen Z, Fasouli ES, Prigmore E, Kleshchevnikov V, Pett JP, Li T, Lawrence JEG, Perera S, Prete M, Huang N, Guo Q, Zeng X, Yang L, Polański K, Chipampe NJ, Dabrowska M, Li X, Bayraktar OA, Patel M, Kumasaka N, Mahbubani KT, Xiang AP, Meyer KB, Saeb-Parsy K, Teichmann SA, Zhang H. Human skeletal muscle aging atlas. NATURE AGING 2024; 4:727-744. [PMID: 38622407 PMCID: PMC11108788 DOI: 10.1038/s43587-024-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species.
Collapse
Affiliation(s)
- Veronika R Kedlian
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tianliang Liu
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Zhuojian Shen
- Department of Thoracic Surgery, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Eirini S Fasouli
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Jan Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - John E G Lawrence
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Qin Guo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinrui Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lu Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krzysztof Polański
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Nana-Jane Chipampe
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Monika Dabrowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xiaobo Li
- Core Facilities for Medical Science, Sun Yat-sen University, Guangzhou, China
| | - Omer Ali Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Minal Patel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Natsuhiko Kumasaka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, UK
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Andy Peng Xiang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK.
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
31
|
von Saucken VE, Windner SE, Baylies MK. Postsynaptic BMP signaling regulates myonuclear properties in Drosophila larval muscles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588944. [PMID: 38645063 PMCID: PMC11030338 DOI: 10.1101/2024.04.10.588944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The syncytial mammalian muscle fiber contains a heterogeneous population of (myo)nuclei. At the neuromuscular junction (NMJ), myonuclei have specialized positioning and gene expression. However, it remains unclear how myonuclei are recruited and what regulates myonuclear output at the NMJ. Here, we identify specific properties of myonuclei located near the Drosophila larval NMJ. These synaptic myonuclei have increased size in relation to their surrounding cytoplasmic domain (scaling), increased DNA content (ploidy), and increased levels of transcription factor pMad, a readout for BMP signaling activity. Our genetic manipulations show local BMP signaling affects muscle size, nuclear size, ploidy, and NMJ size and function. In support, RNA sequencing analysis reveals that pMad regulates genes involved in muscle growth, ploidy (i.e., E2f1), and neurotransmission. Our data suggest that muscle BMP signaling instructs synaptic myonuclear output that then positively shapes the NMJ synapse. This study deepens our understanding of how myonuclear heterogeneity supports local signaling demands to fine tune cellular function and NMJ activity.
Collapse
Affiliation(s)
- Victoria E. von Saucken
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
- Weill Cornell-Rockefeller-Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065 USA
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065 USA
| | - Stefanie E. Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Mary K. Baylies
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| |
Collapse
|
32
|
Wang L, Zhou Y, Wang Y, Shan T. Integrative cross-species analysis reveals conserved and unique signatures in fatty skeletal muscles. Sci Data 2024; 11:290. [PMID: 38472209 DOI: 10.1038/s41597-024-03114-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Fat infiltration in skeletal muscle is now recognized as a standard feature of aging and is directly related to the decline in muscle function. However, there is still a limited systematic integration and exploration of the mechanisms underlying the occurrence of myosteatosis in aging across species. Here, we re-analyzed bulk RNA-seq datasets to investigate the association between fat infiltration in skeletal muscle and aging. Our integrated analysis of single-nucleus transcriptomics in aged humans and Laiwu pigs with high intramuscular fat content, identified species-preference subclusters and revealed core gene programs associated with myosteatosis. Furthermore, we found that fibro/adipogenic progenitors (FAPs) had potential capacity of differentiating into PDE4D+/PDE7B+ preadipocytes across species. Additionally, cell-cell communication analysis revealed that FAPs may be associated with other adipogenic potential clusters via the COL4A2 and COL6A3 pathways. Our study elucidates the correlation mechanism between aging and fat infiltration in skeletal muscle, and these consensus signatures in both humans and pigs may contribute to increasing reproducibility and reliability in future studies involving in the field of muscle research.
Collapse
Affiliation(s)
- Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yanbing Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China.
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
33
|
Darmasaputra GS, van Rijnberk LM, Galli M. Functional consequences of somatic polyploidy in development. Development 2024; 151:dev202392. [PMID: 38415794 PMCID: PMC10946441 DOI: 10.1242/dev.202392] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Polyploid cells contain multiple genome copies and arise in many animal tissues as a regulated part of development. However, polyploid cells can also arise due to cell division failure, DNA damage or tissue damage. Although polyploidization is crucial for the integrity and function of many tissues, the cellular and tissue-wide consequences of polyploidy can be very diverse. Nonetheless, many polyploid cell types and tissues share a remarkable similarity in function, providing important information about the possible contribution of polyploidy to cell and tissue function. Here, we review studies on polyploid cells in development, underlining parallel functions between different polyploid cell types, as well as differences between developmentally-programmed and stress-induced polyploidy.
Collapse
Affiliation(s)
- Gabriella S. Darmasaputra
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| | - Lotte M. van Rijnberk
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| | - Matilde Galli
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, the Netherlands
| |
Collapse
|
34
|
Hsu JE, Ruiz L, Hwang Y, Guzman S, Cho CS, Cheng W, Si Y, Macpherson P, Schrank M, Jun G, Kang HM, Kim M, Brooks S, Lee JH. High-Resolution Spatial Transcriptomic Atlas of Mouse Soleus Muscle: Unveiling Single Cell and Subcellular Heterogeneity in Health and Denervation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582103. [PMID: 38464282 PMCID: PMC10925160 DOI: 10.1101/2024.02.26.582103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Skeletal muscle is essential for both movement and metabolic processes, characterized by a complex and ordered structure. Despite its importance, a detailed spatial map of gene expression within muscle tissue has been challenging to achieve due to the limitations of existing technologies, which struggle to provide high-resolution views. In this study, we leverage the Seq-Scope technique, an innovative method that allows for the observation of the entire transcriptome at an unprecedented submicron spatial resolution. By applying this technique to the mouse soleus muscle, we analyze and compare the gene expression profiles in both healthy conditions and following denervation, a process that mimics aspects of muscle aging. Our approach reveals detailed characteristics of muscle fibers, other cell types present within the muscle, and specific subcellular structures such as the postsynaptic nuclei at neuromuscular junctions, hybrid muscle fibers, and areas of localized expression of genes responsive to muscle injury, along with their histological context. The findings of this research significantly enhance our understanding of the diversity within the muscle cell transcriptome and its variation in response to denervation, a key factor in the decline of muscle function with age. This breakthrough in spatial transcriptomics not only deepens our knowledge of muscle biology but also sets the stage for the development of new therapeutic strategies aimed at mitigating the effects of aging on muscle health, thereby offering a more comprehensive insight into the mechanisms of muscle maintenance and degeneration in the context of aging and disease.
Collapse
Affiliation(s)
- Jer-En Hsu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lloyd Ruiz
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Yongha Hwang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Space Planning and Analysis, University of Michigan, Ann Arbor, MI, USA
| | - Steve Guzman
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Chun-Seok Cho
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Weiqiu Cheng
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Yichen Si
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Peter Macpherson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Mitchell Schrank
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Goo Jun
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hyun-Min Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Susan Brooks
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
35
|
Chrysostomou E, Mourikis P. The extracellular matrix niche of muscle stem cells. Curr Top Dev Biol 2024; 158:123-150. [PMID: 38670702 DOI: 10.1016/bs.ctdb.2024.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Preserving the potency of stem cells in adult tissues is very demanding and relies on the concerted action of various cellular and non-cellular elements in a precise stoichiometry. This balanced microenvironment is found in specific anatomical "pockets" within the tissue, known as the stem cell niche. In this review, we explore the interplay between stem cells and their niches, with a primary focus on skeletal muscle stem cells and the extracellular matrix (ECM). Quiescent muscle stem cells, known as satellite cells are active producers of a diverse array of ECM molecules, encompassing major constituents like collagens, laminins, and integrins, some of which are explored in this review. The conventional perception of ECM as merely a structural scaffold is evolving. Collagens can directly interact as ligands with receptors on satellite cells, while other ECM proteins have the capacity to sequester growth factors and regulate their release, especially relevant during satellite cell turnover in homeostasis or activation upon injury. Additionally, we explore an evolutionary perspective on the ECM across a range of multicellular organisms and discuss a model wherein satellite cells are self-sustained by generating their own niche. Considering the prevalence of ECM proteins in the connective tissue of various organs it is not surprising that mutations in ECM genes have pathological implications, including in muscle, where they can lead to myopathies. However, the particular role of certain disease-related ECM proteins in stem cell maintenance highlights the potential contribution of stem cell deregulation to the progression of these disorders.
Collapse
Affiliation(s)
- Eleni Chrysostomou
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), Créteil, France
| | - Philippos Mourikis
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), Créteil, France.
| |
Collapse
|
36
|
Eguchi T, Tezuka T, Watanabe Y, Inoue-Yamauchi A, Sagara H, Ozawa M, Yamanashi Y. Calcium-binding protein 7 expressed in muscle negatively regulates age-related degeneration of neuromuscular junctions in mice. iScience 2024; 27:108997. [PMID: 38327785 PMCID: PMC10847746 DOI: 10.1016/j.isci.2024.108997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/05/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
The neuromuscular junction (NMJ) forms centrally in myotubes and, as the only synapse between motor neuron and myotube, are indispensable for motor activity. The midmuscle formation of NMJs, including midmuscle-restricted expression of NMJ-related genes, is governed by the muscle-specific kinase (MuSK). However, mechanisms underlying MuSK-mediated signaling are unclear. Here, we find that the Calcium-binding protein 7 (Cabp7) gene shows midmuscle-restricted expression, and muscle-specific depletion of Cabp7 in mice accelerated age-related NMJ degeneration, muscle weakness/atrophy, and motor dysfunction. Surprisingly, forced expression in muscle of CIP, an inhibitory peptide of the negative regulator of NMJ formation cyclin-dependent kinase 5 (Cdk5), restored NMJ integrity and muscle strength, and healed muscle atrophy in muscle-specific Cabp7-deficient mice, which showed increased muscle expression of the Cdk5 activator p25. These findings together demonstrate that MuSK-mediated signaling induces muscle expression of Cabp7, which suppresses age-related NMJ degeneration likely by attenuating p25 expression, providing insights into prophylactic/therapeutic intervention against age-related motor dysfunction.
Collapse
Affiliation(s)
- Takahiro Eguchi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tohru Tezuka
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yuji Watanabe
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Akane Inoue-Yamauchi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Hiroshi Sagara
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Manabu Ozawa
- Laboratory of Reproductive Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Core Laboratory for Developing Advanced Animal Models, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yuji Yamanashi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
37
|
Girolamo DD, Benavente-Diaz M, Murolo M, Grimaldi A, Lopes PT, Evano B, Kuriki M, Gioftsidi S, Laville V, Tinevez JY, Letort G, Mella S, Tajbakhsh S, Comai G. Extraocular muscle stem cells exhibit distinct cellular properties associated with non-muscle molecular signatures. Development 2024; 151:dev202144. [PMID: 38240380 DOI: 10.1242/dev.202144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/27/2023] [Indexed: 02/22/2024]
Abstract
Skeletal muscle stem cells (MuSCs) are recognised as functionally heterogeneous. Cranial MuSCs are reported to have greater proliferative and regenerative capacity when compared with those in the limb. A comprehensive understanding of the mechanisms underlying this functional heterogeneity is lacking. Here, we have used clonal analysis, live imaging and single cell transcriptomic analysis to identify crucial features that distinguish extraocular muscle (EOM) from limb muscle stem cell populations. A MyogeninntdTom reporter showed that the increased proliferation capacity of EOM MuSCs correlates with deferred differentiation and lower expression of the myogenic commitment gene Myod. Unexpectedly, EOM MuSCs activated in vitro expressed a large array of extracellular matrix components typical of mesenchymal non-muscle cells. Computational analysis underscored a distinct co-regulatory module, which is absent in limb MuSCs, as driver of these features. The EOM transcription factor network, with Foxc1 as key player, appears to be hardwired to EOM identity as it persists during growth, disease and in vitro after several passages. Our findings shed light on how high-performing MuSCs regulate myogenic commitment by remodelling their local environment and adopting properties not generally associated with myogenic cells.
Collapse
Affiliation(s)
- Daniela Di Girolamo
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Maria Benavente-Diaz
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Sorbonne Universités, Complexité du Vivant, F-75005 Paris, France
| | - Melania Murolo
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Alexandre Grimaldi
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Sorbonne Universités, Complexité du Vivant, F-75005 Paris, France
| | - Priscilla Thomas Lopes
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Brendan Evano
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Mao Kuriki
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Stamatia Gioftsidi
- Université Paris-Est, 77420 Champs-sur- Marne, France
- Freie Universität Berlin, 14195 Berlin, Germany
- Inserm, IMRB U955-E10, 94000 Créteil, France
| | - Vincent Laville
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
| | - Jean-Yves Tinevez
- Institut Pasteur, Université Paris Cité, Image Analysis Hub, 75015 Paris, France
| | - Gaëlle Letort
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Sebastian Mella
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Glenda Comai
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| |
Collapse
|
38
|
Tong S, Sun Y, Kuang B, Wang M, Chen Z, Zhang W, Chen J. A Comprehensive Review of Muscle-Tendon Junction: Structure, Function, Injury and Repair. Biomedicines 2024; 12:423. [PMID: 38398025 PMCID: PMC10886980 DOI: 10.3390/biomedicines12020423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The muscle-tendon junction (MTJ) is a highly specific tissue interface where the muscle's fascia intersects with the extracellular matrix of the tendon. The MTJ functions as the particular structure facilitating the transmission of force from contractive muscle fibers to the skeletal system, enabling movement. Considering that the MTJ is continuously exposed to constant mechanical forces during physical activity, it is susceptible to injuries. Ruptures at the MTJ often accompany damage to both tendon and muscle tissues. In this review, we attempt to provide a precise definition of the MTJ, describe its subtle structure in detail, and introduce therapeutic approaches related to MTJ tissue engineering. We hope that our detailed illustration of the MTJ and summary of the representative research achievements will help researchers gain a deeper understanding of the MTJ and inspire fresh insights and breakthroughs for future research.
Collapse
Affiliation(s)
- Siqi Tong
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Yuzhi Sun
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Baian Kuang
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Mingyue Wang
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| |
Collapse
|
39
|
Cisterna B, Malatesta M. Molecular and Structural Alterations of Skeletal Muscle Tissue Nuclei during Aging. Int J Mol Sci 2024; 25:1833. [PMID: 38339110 PMCID: PMC10855217 DOI: 10.3390/ijms25031833] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Aging is accompanied by a progressive loss of skeletal muscle mass and strength. The mechanisms underlying this phenomenon are certainly multifactorial and still remain to be fully elucidated. Changes in the cell nucleus structure and function have been considered among the possible contributing causes. This review offers an overview of the current knowledge on skeletal muscle nuclei in aging, focusing on the impairment of nuclear pathways potentially involved in age-related muscle decline. In skeletal muscle two types of cells are present: fiber cells, constituting the contractile muscle mass and containing hundreds of myonuclei, and the satellite cells, i.e., the myogenic mononuclear stem cells occurring at the periphery of the fibers and responsible for muscle growth and repair. Research conducted on different experimental models and with different methodological approaches demonstrated that both the myonuclei and satellite cell nuclei of aged skeletal muscles undergo several structural and molecular alterations, affecting chromatin organization, gene expression, and transcriptional and post-transcriptional activities. These alterations play a key role in the impairment of muscle fiber homeostasis and regeneration, thus contributing to the age-related decrease in skeletal muscle mass and function.
Collapse
Affiliation(s)
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy;
| |
Collapse
|
40
|
Xiao W, Jiang N, Ji Z, Ni M, Zhang Z, Zhao Q, Huang R, Li P, Hou L. Single-Cell RNA Sequencing Reveals the Cellular Landscape of Longissimus Dorsi in a Newborn Suhuai Pig. Int J Mol Sci 2024; 25:1204. [PMID: 38256277 PMCID: PMC10816681 DOI: 10.3390/ijms25021204] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
The introduction of single-cell RNA sequencing (scRNA-seq) technology has spurred additional advancements in analyzing the cellular composition of tissues. The longissimus dorsi (LD) in pigs serves as the primary skeletal muscle for studying meat quality in the pig industry. However, the single-cell profile of porcine LD is still in its infancy stage. In this study, we profiled the transcriptomes of 16,018 cells in the LD of a newborn Suhuai pig at single-cell resolution. Subsequently, we constructed a cellular atlas of the LD, identifying 11 distinct cell populations, including endothelial cells (24.39%), myotubes (18.82%), fibro-adipogenic progenitors (FAPs, 18.11%), satellite cells (16.74%), myoblasts (3.99%), myocytes (5.74%), Schwann cells (3.81%), smooth muscle cells (3.22%), dendritic cells (2.99%), pericytes (1.86%), and neutrophils (0.33%). CellChat was employed to deduce the cell-cell interactions by evaluating the gene expression of receptor-ligand pairs across different cell types. The results show that FAPs and pericytes are the primary signal contributors in LD. In addition, we delineated the developmental trajectory of myogenic cells and examined alterations in the expression of various marker genes and molecular events throughout various stages of differentiation. Moreover, we found that FAPs can be divided into three subclusters (NR2F2-FAPs, LPL-FAPs, and TNMD-FAPs) according to their biological functions, suggesting that the FAPs could be associated with the differentiation of tendon cell. Taken together, we constructed the cellular atlas and cell communication network in LD of a newborn Suhuai pig, and analyzed the developmental trajectory of myogenic cells and the heterogeneity of FAPs subpopulation cells. This enhances our comprehension of the molecular features involved in skeletal muscle development and the meat quality control in pigs.
Collapse
Affiliation(s)
- Wei Xiao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Nengjing Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhengyu Ji
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Mengru Ni
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhaobo Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Qingbo Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
- Huai’an Academy, Nanjing Agricultural University, Huai’an 223001, China
| | - Ruihua Huang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
- Huai’an Academy, Nanjing Agricultural University, Huai’an 223001, China
| | - Pinghua Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
- Huai’an Academy, Nanjing Agricultural University, Huai’an 223001, China
| | - Liming Hou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (W.X.); (N.J.); (Z.J.); (M.N.); (Z.Z.); (Q.Z.); (R.H.); (P.L.)
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Pig Genetic Resources Evaluation and Utilization (Nanjing) of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210095, China
- Huai’an Academy, Nanjing Agricultural University, Huai’an 223001, China
| |
Collapse
|
41
|
Banks RW. There and back again: 50 years of wandering through terra incognita fusorum. Exp Physiol 2024; 109:6-16. [PMID: 36628601 PMCID: PMC10988739 DOI: 10.1113/ep090760] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/12/2022] [Indexed: 01/12/2023]
Abstract
This paper is in two parts: 'There', which is a review of some of the major advances in the study of spindle structure and function during the past 50 years, serving as an introduction to the symposium entitled 'Mechanotransduction, Muscle Spindles and Proprioception' held in Munich in July 2022; and 'And Back Again', presenting new quantitative morphological results on the equatorial nuclei of intrafusal muscle fibres and of the primary sensory ending in relationship to passive stretch of the spindle.
Collapse
Affiliation(s)
- Robert W. Banks
- Department of BiosciencesUniversity of DurhamDurhamUK
- Biophysical Sciences InstituteUniversity of DurhamDurhamUK
| |
Collapse
|
42
|
Kröger S. Experimental Physiology special issue: 'Mechanotransduction, muscle spindles and proprioception'. Exp Physiol 2024; 109:1-5. [PMID: 38160398 PMCID: PMC10988673 DOI: 10.1113/ep091431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Affiliation(s)
- Stephan Kröger
- Department of Physiological Genomics, Biomedical CenterLudwig‐Maximilians‐UniversitätPlanegg‐MartinsriedGermany
| |
Collapse
|
43
|
Bazin-Gélis M, Eleftheriou E, Zangarelli C, Lelandais G, Sperling L, Arnaiz O, Bétermier M. Inter-generational nuclear crosstalk links the control of gene expression to programmed genome rearrangement during the Paramecium sexual cycle. Nucleic Acids Res 2023; 51:12337-12351. [PMID: 37953377 PMCID: PMC10711438 DOI: 10.1093/nar/gkad1006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023] Open
Abstract
Multinucleate cells are found in many eukaryotes, but how multiple nuclei coordinate their functions is still poorly understood. In the cytoplasm of the ciliate Paramecium tetraurelia, two micronuclei (MIC) serving sexual reproduction coexist with a somatic macronucleus (MAC) dedicated to gene expression. During sexual processes, the MAC is progressively destroyed while still ensuring transcription, and new MACs develop from copies of the zygotic MIC. Several gene clusters are successively induced and switched off before vegetative growth resumes. Concomitantly, programmed genome rearrangement (PGR) removes transposons and their relics from the new MACs. Development of the new MACs is controlled by the old MAC, since the latter expresses genes involved in PGR, including the PGM gene encoding the essential PiggyMac endonuclease that cleaves the ends of eliminated sequences. Using RNA deep sequencing and transcriptome analysis, we show that impairing PGR upregulates key known PGR genes, together with ∼600 other genes possibly also involved in PGR. Among these genes, 42% are no longer induced when no new MACs are formed, including 180 genes that are co-expressed with PGM under all tested conditions. We propose that bi-directional crosstalk between the two coexisting generations of MACs links gene expression to the progression of MAC development.
Collapse
Affiliation(s)
- Mélanie Bazin-Gélis
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Evangelia Eleftheriou
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
- Institut Pasteur, Université Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Coralie Zangarelli
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Gaëlle Lelandais
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Linda Sperling
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Olivier Arnaiz
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Mireille Bétermier
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| |
Collapse
|
44
|
Ismaeel A, Thomas NT, McCashland M, Vechetti IJ, Edman S, Lanner JT, Figueiredo VC, Fry CS, McCarthy JJ, Wen Y, Murach KA, von Walden F. Coordinated Regulation of Myonuclear DNA Methylation, mRNA, and miRNA Levels Associates With the Metabolic Response to Rapid Synergist Ablation-Induced Skeletal Muscle Hypertrophy in Female Mice. FUNCTION 2023; 5:zqad062. [PMID: 38020067 PMCID: PMC10666992 DOI: 10.1093/function/zqad062] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/16/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
The central dogma of molecular biology dictates the general flow of molecular information from DNA that leads to a functional cellular outcome. In skeletal muscle fibers, the extent to which global myonuclear transcriptional alterations, accounting for epigenetic and post-transcriptional influences, contribute to an adaptive stress response is not clearly defined. In this investigation, we leveraged an integrated analysis of the myonucleus-specific DNA methylome and transcriptome, as well as myonuclear small RNA profiling to molecularly define the early phase of skeletal muscle fiber hypertrophy. The analysis of myonucleus-specific mature microRNA and other small RNA species provides new directions for exploring muscle adaptation and complemented the methylation and transcriptional information. Our integrated multi-omics interrogation revealed a coordinated myonuclear molecular landscape during muscle loading that coincides with an acute and rapid reduction of oxidative metabolism. This response may favor a biosynthesis-oriented metabolic program that supports rapid hypertrophic growth.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40508, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Mariah McCashland
- Department of Nutrition and Health Sciences, University of Nebraska–Lincoln, Lincoln, NE 68583, USA
| | - Ivan J Vechetti
- Department of Nutrition and Health Sciences, University of Nebraska–Lincoln, Lincoln, NE 68583, USA
| | - Sebastian Edman
- Department of Women’s and Children’s Health, Karolinska Institutet, Solna 17177, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna 17177, Sweden
| | - Vandré C Figueiredo
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40508, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40508, USA
- Center for Muscle Biology, University of Kentucky, Lexington, KY 40536, USA
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, AR 72701, USA
| | - Ferdinand von Walden
- Department of Women’s and Children’s Health, Karolinska Institutet, Solna 17177, Sweden
| |
Collapse
|
45
|
Nelke C, Schroeter CB, Theissen L, Preusse C, Pawlitzki M, Räuber S, Dobelmann V, Cengiz D, Kleefeld F, Roos A, Schoser B, Brunn A, Neuen-Jacob E, Zschüntzsch J, Meuth SG, Stenzel W, Ruck T. Senescent fibro-adipogenic progenitors are potential drivers of pathology in inclusion body myositis. Acta Neuropathol 2023; 146:725-745. [PMID: 37773216 PMCID: PMC10564677 DOI: 10.1007/s00401-023-02637-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Inclusion body myositis (IBM) is unique across the spectrum of idiopathic inflammatory myopathies (IIM) due to its distinct clinical presentation and refractoriness to current treatment approaches. One explanation for this resistance may be the engagement of cell-autonomous mechanisms that sustain or promote disease progression of IBM independent of inflammatory activity. In this study, we focused on senescence of tissue-resident cells as potential driver of disease. For this purpose, we compared IBM patients to non-diseased controls and immune-mediated necrotizing myopathy patients. Histopathological analysis suggested that cellular senescence is a prominent feature of IBM, primarily affecting non-myogenic cells. In-depth analysis by single nuclei RNA sequencing allowed for the deconvolution and study of muscle-resident cell populations. Among these, we identified a specific cluster of fibro-adipogenic progenitors (FAPs) that demonstrated key hallmarks of senescence, including a pro-inflammatory secretome, expression of p21, increased β-galactosidase activity, and engagement of senescence pathways. FAP function is required for muscle cell health with changes to their phenotype potentially proving detrimental. In this respect, the transcriptomic landscape of IBM was also characterized by changes to the myogenic compartment demonstrating a pronounced loss of type 2A myofibers and a rarefication of acetylcholine receptor expressing myofibers. IBM muscle cells also engaged a specific pro-inflammatory phenotype defined by intracellular complement activity and the expression of immunogenic surface molecules. Skeletal muscle cell dysfunction may be linked to FAP senescence by a change in the collagen composition of the latter. Senescent FAPs lose collagen type XV expression, which is required to support myofibers' structural integrity and neuromuscular junction formation in vitro. Taken together, this study demonstrates an altered phenotypical landscape of muscle-resident cells and that FAPs, and not myofibers, are the primary senescent cell type in IBM.
Collapse
Affiliation(s)
- Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Lukas Theissen
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Corinna Preusse
- Department of Neuropathology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Saskia Räuber
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Vera Dobelmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Derya Cengiz
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Felix Kleefeld
- Department of Neurology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Andreas Roos
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, Centre for Neuromuscular Disorders in Children, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Benedikt Schoser
- Friedrich Baur Institute at the Department of Neurology, LMU University Hospital, LMU Munich, 80336, Munich, Germany
| | - Anna Brunn
- Institute of Neuropathology, Heinrich Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Eva Neuen-Jacob
- Institute of Neuropathology, Heinrich Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité-University Medicine Berlin, Bonhoefferweg 3, 10117, Berlin, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| |
Collapse
|
46
|
Ruiz LP, Macpherson PC, Brooks SV. Maintenance of subsynaptic myonuclei number is not driven by neural input. Front Physiol 2023; 14:1266950. [PMID: 37822678 PMCID: PMC10562629 DOI: 10.3389/fphys.2023.1266950] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
The development and maintenance of neuromuscular junctions (NMJ) are supported by a specialized population of myonuclei that are referred to as the subsynaptic myonuclei (SSM). The relationship between the number of SSM and the integrity of the NMJ as well as the impact of a loss of innervation on SSM remain unclear. This study aimed to clarify these associations by simultaneously analyzing SSM counts and NMJ innervation status in three distinct mouse models of acute and chronic NMJ disruption. SSM were identified using fluorescent immunohistochemistry for Nesprin1 expression, which is highly enriched in SSM, along with anatomical location beneath the muscle fiber motor endplate. Acute denervation, induced by surgical nerve transection, did not affect SSM number after 7 days. Additionally, no significant changes in SSM number were observed during normal aging or in mice with chronic oxidative stress (Sod1 -/-). Both aging WT mice and Sod1 -/- mice accumulated degenerating and denervated NMJ in skeletal muscle, but there was no correlation between innervation status of a given NMJ and SSM number in aged or Sod1 -/- mice. These findings challenge the notion that a loss of SSM is a primary driver of NMJ degradation and leave open questions of the mechanisms that regulate SSM number as well as the physiological significance of the precise SSM number. Further investigations are required to define other properties of the SSM, such as transcriptional profiles and structural integrity, to better understand their role in NMJ maintenance.
Collapse
Affiliation(s)
- Lloyd P. Ruiz
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Peter C. Macpherson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Susan V. Brooks
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
47
|
Oprescu SN, Baumann N, Chen X, Sun Q, Zhao Y, Yue F, Wang H, Kuang S. Sox11 is enriched in myogenic progenitors but dispensable for development and regeneration of the skeletal muscle. Skelet Muscle 2023; 13:15. [PMID: 37705115 PMCID: PMC10498607 DOI: 10.1186/s13395-023-00324-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
Transcription factors (TFs) play key roles in regulating differentiation and function of stem cells, including muscle satellite cells (MuSCs), a resident stem cell population responsible for postnatal regeneration of the skeletal muscle. Sox11 belongs to the Sry-related HMG-box (SOX) family of TFs that play diverse roles in stem cell behavior and tissue specification. Analysis of single-cell RNA-sequencing (scRNA-seq) datasets identify a specific enrichment of Sox11 mRNA in differentiating but not quiescent MuSCs. Consistent with the scRNA-seq data, Sox11 levels increase during differentiation of murine primary myoblasts in vitro. scRNA-seq data comparing muscle regeneration in young and old mice further demonstrate that Sox11 expression is reduced in aged MuSCs. Age-related decline of Sox11 expression is associated with reduced chromatin contacts within the topologically associating domains. Unexpectedly, Myod1Cre-driven deletion of Sox11 in embryonic myoblasts has no effects on muscle development and growth, resulting in apparently healthy muscles that regenerate normally. Pax7CreER- or Rosa26CreER- driven (MuSC-specific or global) deletion of Sox11 in adult mice similarly has no effects on MuSC differentiation or muscle regeneration. These results identify Sox11 as a novel myogenic differentiation marker with reduced expression in quiescent and aged MuSCs, but the specific function of Sox11 in myogenesis remains to be elucidated.
Collapse
Affiliation(s)
- Stephanie N Oprescu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Nick Baumann
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiyue Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Qiang Sun
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong, China
| | - Yu Zhao
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong, China
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Huating Wang
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong, China
| | - Shihuan Kuang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
48
|
Smith JAB, Murach KA, Dyar KA, Zierath JR. Exercise metabolism and adaptation in skeletal muscle. Nat Rev Mol Cell Biol 2023; 24:607-632. [PMID: 37225892 PMCID: PMC10527431 DOI: 10.1038/s41580-023-00606-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/26/2023]
Abstract
Viewing metabolism through the lens of exercise biology has proven an accessible and practical strategy to gain new insights into local and systemic metabolic regulation. Recent methodological developments have advanced understanding of the central role of skeletal muscle in many exercise-associated health benefits and have uncovered the molecular underpinnings driving adaptive responses to training regimens. In this Review, we provide a contemporary view of the metabolic flexibility and functional plasticity of skeletal muscle in response to exercise. First, we provide background on the macrostructure and ultrastructure of skeletal muscle fibres, highlighting the current understanding of sarcomeric networks and mitochondrial subpopulations. Next, we discuss acute exercise skeletal muscle metabolism and the signalling, transcriptional and epigenetic regulation of adaptations to exercise training. We address knowledge gaps throughout and propose future directions for the field. This Review contextualizes recent research of skeletal muscle exercise metabolism, framing further advances and translation into practice.
Collapse
Affiliation(s)
- Jonathon A B Smith
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kevin A Murach
- Molecular Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Kenneth A Dyar
- Metabolic Physiology, Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
49
|
Corvelyn M, Meirlevede J, Deschrevel J, Huyghe E, De Wachter E, Gayan-Ramirez G, Sampaolesi M, Van Campenhout A, Desloovere K, Costamagna D. Ex vivo adult stem cell characterization from multiple muscles in ambulatory children with cerebral palsy during early development of contractures. Differentiation 2023; 133:25-39. [PMID: 37451110 DOI: 10.1016/j.diff.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 05/25/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
Cerebral palsy (CP) is one of the most common conditions leading to lifelong childhood physical disability. Literature reported previously altered muscle properties such as lower number of satellite cells (SCs), with altered fusion capacity. However, these observations highly vary among studies, possibly due to heterogeneity in patient population, lack of appropriate control data, methodology and different assessed muscle. In this study we aimed to strengthen previous observations and to understand the heterogeneity of CP muscle pathology. Myogenic differentiation of SCs from the Medial Gastrocnemius (MG) muscle of patients with CP (n = 16, 3-9 years old) showed higher fusion capacity compared to age-matched typically developing children (TD, n = 13). Furthermore, we uniquely assessed cells of two different lower limb muscles and showed a decreased myogenic potency in cells from the Semitendinosus (ST) compared to the MG (TD: n = 3, CP: n = 6). Longitudinal assessments, one year after the first botulinum toxin treatment, showed slightly reduced SC representations and lower fusion capacity (n = 4). Finally, we proved the robustness of our data, by assessing in parallel the myogenic capacity of two samples from the same TD muscle. In conclusion, these data confirmed previous findings of increased SC fusion capacity from MG muscle of young patients with CP compared to age-matched TD. Further elaboration is reported on potential factors contributing to heterogeneity, such as assessed muscle, CP progression and reliability of primary outcome parameters.
Collapse
Affiliation(s)
- M Corvelyn
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium
| | - J Meirlevede
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium
| | - J Deschrevel
- Laboratory of Respiratory Diseases and Thoracic Surgery, Dept. of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - E Huyghe
- Research Group for Neurorehabilitation, Dept. of Rehabilitation Sciences, KU Leuven, Belgium
| | - E De Wachter
- Dept. of Orthopaedic Surgery, University Hospitals Leuven, Belgium
| | - G Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery, Dept. of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - M Sampaolesi
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium
| | - A Van Campenhout
- Dept. of Orthopaedic Surgery, University Hospitals Leuven, Belgium; Dept. of Development and Regeneration, KU Leuven, Belgium
| | - K Desloovere
- Research Group for Neurorehabilitation, Dept. of Rehabilitation Sciences, KU Leuven, Belgium.
| | - D Costamagna
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium; Research Group for Neurorehabilitation, Dept. of Rehabilitation Sciences, KU Leuven, Belgium.
| |
Collapse
|
50
|
Sun C, Swoboda CO, Petrany MJ, Parameswaran S, VonHandorf A, Weirauch MT, Lepper C, Millay DP. Lineage tracing of newly accrued nuclei in skeletal myofibers uncovers distinct transcripts and interplay between nuclear populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554609. [PMID: 37662191 PMCID: PMC10473681 DOI: 10.1101/2023.08.24.554609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Multinucleated skeletal muscle cells have an obligatory need to acquire additional nuclei through fusion with activated skeletal muscle stem cells when responding to both developmental and adaptive growth stimuli. A fundamental question in skeletal muscle biology has been the reason underlying this need for new nuclei in syncytial cells that already harbor hundreds of nuclei. To begin to answer this long-standing question, we utilized nuclear RNA-sequencing approaches and developed a lineage tracing strategy capable of defining the transcriptional state of recently fused nuclei and distinguishing this state from that of pre-existing nuclei. Our findings reveal the presence of conserved markers of newly fused nuclei both during development and after a hypertrophic stimulus in the adult. However, newly fused nuclei also exhibit divergent gene expression that is determined by the myogenic environment to which they fuse. Moreover, accrual of new nuclei through fusion is required for nuclei already resident in adult myofibers to mount a normal transcriptional response to a load-inducing stimulus. We propose a model of mutual regulation in the control of skeletal muscle development and adaptations, where newly fused and pre-existing myonuclear populations influence each other to maintain optimal functional growth.
Collapse
Affiliation(s)
- Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O. Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Michael J. Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Sreeja Parameswaran
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew VonHandorf
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T. Weirauch
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Center for Autoimmune Genetics and Etiology (CAGE), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Douglas P. Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|